Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

new england

The
journal of medicine
established in 1812 August 5, 2021 vol. 385  no. 6

CRISPR-Cas9 In Vivo Gene Editing for Transthyretin Amyloidosis


Julian D. Gillmore, M.D., Ph.D., Ed Gane, M.B., Ch.B., Jorg Taubel, M.D., Justin Kao, M.B., Ch.B.,
Marianna Fontana, M.D., Ph.D., Michael L. Maitland, M.D., Ph.D., Jessica Seitzer, B.S., Daniel O’Connell, Ph.D.,
Kathryn R. Walsh, Ph.D., Kristy Wood, Ph.D., Jonathan Phillips, Ph.D., Yuanxin Xu, M.D., Ph.D., Adam Amaral, B.A.,
Adam P. Boyd, Ph.D., Jeffrey E. Cehelsky, M.B.A., Mark D. McKee, M.D., Andrew Schiermeier, Ph.D.,
Olivier Harari, M.B., B.Chir., Ph.D., Andrew Murphy, Ph.D., Christos A. Kyratsous, Ph.D., Brian Zambrowicz, Ph.D.,
Randy Soltys, Ph.D., David E. Gutstein, M.D., John Leonard, M.D., Laura Sepp‑Lorenzino, Ph.D.,
and David Lebwohl, M.D.​​

a bs t r ac t

BACKGROUND
Transthyretin amyloidosis, also called ATTR amyloidosis, is a life-threatening From the National Amyloidosis Centre,
Division of Medicine, University College
disease characterized by progressive accumulation of misfolded transthyretin
London, Royal Free Hospital (J.D.G.,
(TTR) protein in tissues, predominantly the nerves and heart. NTLA-2001 is an in M.F.) and Richmond Pharmacology, St.
vivo gene-editing therapeutic agent that is designed to treat ATTR amyloidosis by George’s University of London (J.T.) —
both in London; New Zealand Clinical
reducing the concentration of TTR in serum. It is based on the clustered regu-
Research (E.G.), University of Auckland
larly interspaced short palindromic repeats and associated Cas9 endonuclease (E.G.), and the Department of Neurolo-
(CRISPR-Cas9) system and comprises a lipid nanoparticle encapsulating messenger gy, Auckland City Hospital (J.K.) — all in
Auckland, New Zealand; Intellia Thera-
RNA for Cas9 protein and a single guide RNA targeting TTR.
peutics, Cambridge, MA (M.L.M., J.S.,
D.O., K.R.W., K.W., J.P., Y.X., A.A., A.P.B.,
METHODS J.E.C., M.D.M., A.S., J.L., L.S.-L., D.L.);
After conducting preclinical in vitro and in vivo studies, we evaluated the safety and Regeneron Pharmaceuticals, Tarry-
and pharmacodynamic effects of single escalating doses of NTLA-2001 in six pa- town, NY (O.H., A.M., C.A.K., B.Z., R.S.,
D.E.G.). Address reprint requests to Prof.
tients with hereditary ATTR amyloidosis with polyneuropathy, three in each of the Gillmore at the Centre for Amyloidosis
two initial dose groups (0.1 mg per kilogram and 0.3 mg per kilogram), within an and Acute Phase Proteins, Division of
ongoing phase 1 clinical study. Medicine, University College London,
Royal Free Hospital, Rowland Hill St.,
London NW3 2PF, United Kingdom, or at
RESULTS
­j​.­gillmore@​­ucl​.­ac​.­uk.
Preclinical studies showed durable knockout of TTR after a single dose. Serial as-
This article was published on June 26,
sessments of safety during the first 28 days after infusion in patients revealed few
2021, at NEJM.org.
adverse events, and those that did occur were mild in grade. Dose-dependent
N Engl J Med 2021;385:493-502.
pharmacodynamic effects were observed. At day 28, the mean reduction from
DOI: 10.1056/NEJMoa2107454
baseline in serum TTR protein concentration was 52% (range, 47 to 56) in the Copyright © 2021 Massachusetts Medical Society.
group that received a dose of 0.1 mg per kilogram and was 87% (range, 80 to 96)
in the group that received a dose of 0.3 mg per kilogram.
CONCLUSIONS
In a small group of patients with hereditary ATTR amyloidosis with polyneuropa-
thy, administration of NTLA-2001 was associated with only mild adverse events
and led to decreases in serum TTR protein concentrations through targeted
knockout of TTR. (Funded by Intellia Therapeutics and Regeneron Pharmaceuti-
cals; ClinicalTrials.gov number, NCT04601051.)

n engl j med 385;6  nejm.org  August 5, 2021 493


The New England Journal of Medicine
Downloaded from nejm.org on July 11, 2023. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

T
ransthyretin amyloidosis, also clustered regularly interspaced short palindrom-
called ATTR amyloidosis, is a progressive ic repeats and associated Cas9 endonuclease
fatal disease characterized by accumula- (CRISPR-Cas9) system to achieve in vivo gene
tion in tissues of amyloid fibrils composed of editing.18-21 As a monogenic disease, ATTR amy-
misfolded transthyretin (TTR) protein.1,2 ATTR loidosis represents an ideal target for the appli-
A Quick Take
is available at amyloidosis may be acquired; referred to as wild- cation of CRISPR-Cas9–mediated in vivo gene
NEJM.org type ATTR amyloidosis, this form of ATTR amy- editing. The limited and specific normal func-
loidosis is an increasingly recognized cause of tion of TTR in thyroxine and vitamin A trans-
cardiomyopathy and heart failure.2 In rarer cases, port22 means that knockdown has only limited
ATTR amyloidosis is hereditary (known as vari- additional physiological effects23; in addition,
ant or hereditary ATTR [hATTR] amyloidosis); circulating TTR is produced almost entirely (>99%)
hATTR amyloidosis can be triggered by more within the liver,2 for which established targeting
than 100 different pathogenic mutations in TTR.3 systems such as lipid nanoparticles are available.
The hereditary form of ATTR amyloidosis is NTLA-2001 is a new CRISPR-Cas9–based in
thought to be present in approximately 50,000 vivo gene-editing therapy, administered by intra-
persons worldwide4,5; it has an autosomal domi- venous infusion, that is intended to edit TTR in
nant pattern of inheritance and a clinical pheno- hepatocytes, leading to a decrease in the produc-
type dominated by amyloid polyneuropathy or tion of both wild-type and mutant TTR after a
cardiomyopathy, with most patients having a single administration. In mouse models and non-
combination of the two.6 After the onset of symp- human primates (cynomolgus monkeys), single
toms, ATTR amyloidosis is progressive, culmi- doses resulted in durable reductions in serum
nating in death within a median of 2 to 6 years TTR protein of 95% or greater24,25 and therefore
after diagnosis in patients with amyloid cardio- provide potentially greater TTR knockdown
myopathy7 and 4 to 17 years after symptom onset than currently available therapies.
in patients with amyloid polyneuropathy in the NTLA-2001 consists of a proprietary lipid
absence of cardiomyopathy.8 nanoparticle (LNP) delivery system with liver
Current therapeutic strategies for ATTR amy- tropism, carrying a single guide RNA (sgRNA)
loidosis rely on reducing ongoing amyloid for- that targets human TTR and a human-codon–
mation through stabilization of the tetrameric optimized mRNA sequence of Streptococcus pyo-
form of TTR (with diflunisal or tafamidis)9,10 or genes Cas9 protein (Fig. 1). LNPs have previously
through inhibition of TTR protein synthesis been used in vivo for liver-targeted delivery of a
(with inotersen or patisiran) by means of degra- variety of therapeutic RNA cargoes, such as small
dation of TTR messenger RNA (mRNA).11,12 Such interfering RNA and mRNA.26,27 For NTLA-2001,
treatments produce symptom relief and functional the LNP formulation was developed for genome
improvement and prolong survival12-14 but are lim- editing with regard to lipid composition and RNA
ited by the requirement for long-term adminis- cargo in order to enable efficient delivery to the
tration to maintain TTR knockdown. In the case liver in a variety of preclinical models. As de-
of patisiran, long-term treatment results in con- scribed previously, on polyethylene glycol–lipid
tinued exposure to premedication with glucocor- diffusion from this LNP, plasma apolipoprotein
ticoids and antihistamines.15 In addition, patients E binds to (opsonizes) the LNP surface in circu-
receiving TTR-stabilizing agents have disease pro- lation; the LNP is then actively endocytosed by
gression.16 Inotersen is associated with serious hepatocytes through the low-density lipoprotein
side effects, including glomerulonephritis and receptor.27 Given that the liver is the almost exclu-
decreased platelet counts.17 More extensive TTR sive site of TTR manufacture,2 this liver-targeting
knockdown is associated with greater improve- delivery system should maximize efficacy while
ment in neuropathic end points in patients with minimizing systemic toxic effects.
hATTR polyneuropathy.12 Enhancements in TTR Here, we report interim data from an ongoing
reduction, including sustained knockdown, may clinical study evaluating single ascending doses
translate into improved outcomes for patients with of NTLA-2001 for TTR editing and knockout in
ATTR amyloidosis. A potential alternative to the treatment of patients with hATTR amyloido-
mRNA targeting-based gene silencing is use of the sis with polyneuropathy.

494 n engl j med 385;6  nejm.org  August 5, 2021

The New England Journal of Medicine


Downloaded from nejm.org on July 11, 2023. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
CRISPR-C as 9 for Tr ansthyretin Amyloidosis

Me thods and the United Kingdom and were analyzed by


the sponsors. A medical writer prepared the first
Preclinical Studies draft of the manuscript, under direction from
An sgRNA targeting the TTR sequence the authors and with funding from the sponsors.
AAAGGCUGCUGAUGACACCU (human genome The authors had access to the data and collabo-
build hg38, chromosome 18: 31592987–31593007) rated in the preparation and revisions of manu-
was selected for efficient knockout and specific- script drafts before and after submission. The
ity after a comprehensive off-target characteriza- investigators and sponsors vouch for the accu-
tion workflow that applied a combination of racy and completeness of the data and for the
both computational modeling and empirical fidelity of the study to the protocol. The protocol
approaches. To select for a high therapeutic in- was reviewed by national and institutional ethics
dex (i.e., the ratio of on-target to off-target edit- and regulatory bodies, including expert commit-
ing), we performed genomewide assays and tar- tees for the assessment of new studies of gene
geted sequencing to identify and verify candidate therapy, such as gene editing (Gene Therapy Ad-
sgRNA off-target sites. The in vitro dose–response visory Committee of the Ministry of Health, New
and gene-editing potency of NTLA-2001 were Zealand, and Gene Therapy Advisory Committee
assessed in primary cell cultures of human he- of the Medicines and Healthcare Products Regu-
patocytes. Genomic loci with the potential for latory Agency, United Kingdom). The study has
off-target editing were found with the use of been conducted in accordance with the Declara-
complementary computational and laboratory- tion of Helsinki and International Council for
based approaches (Cas-OFFinder,28 GUIDE-seq,29 Harmonisation Good Clinical Practice guide-
and SITE-Seq30). Candidate loci were validated lines, and all patients provided written informed
for the detection of off-target insertions and consent.
deletions (indels) with the use of next-generation
sequencing after NTLA-2001 treatment of pri- Clinical Study Design and Eligibility
mary human hepatocytes at concentrations up to We report the results in two initial dose groups
27 times as high as concentrations that achieved from part 1 of a two-part, global, phase 1, open-
greater than 90% reduction in TTR protein (EC90). label, multicenter study. Patients were treated with
Additional details of these studies are provided a single dose of NTLA-2001 consisting of a total
in the Supplementary Appendix, available with RNA dose of 0.1 mg per kilogram of body weight
the full text of this article at NEJM.org. or 0.3 mg per kilogram of body weight adminis-
Transgenic mouse models with either wild- tered intravenously between November 2020 and
type or mutant (p.V50M) human TTR were used April 2021. Key eligibility criteria for part 1 of the
for initial proof-of-mechanism studies of NTLA- study included an age of 18 to 80 years, a diag-
2001 in which we evaluated TTR editing and re- nosis of polyneuropathy due to hATTR amyloi-
ductions in circulating TTR protein levels.24 The dosis (with or without cardiomyopathy), a body
cynomolgus monkey was selected as a relevant weight of 50 to 90 kg at the screening visit, and
nonclinical animal model for the evaluation of a lack of access to approved treatments for ATTR
pharmacologic, toxicologic, pharmacokinetic, amyloidosis. Patients with non-ATTR amyloido-
and metabolic properties, including TTR editing, sis, known leptomeningeal ATTR amyloidosis,
reductions in circulating TTR protein levels, and or a history of receipt of RNA-silencing therapy
serum concentrations of LNP and cargo compo- were excluded. Previous use of TTR stabilizers
nents over time.25 was permitted with a washout period (3 days for
diflunisal).
Clinical Study Oversight
The clinical study is sponsored by Intellia Thera- Clinical Study Treatment
peutics and Regeneron Pharmaceuticals; the spon- Safety studies in cynomolgus monkeys were used
sors designed the protocol, available at NEJM.org. to determine the no-observed-adverse-effect level
Study oversight was provided by an independent (NOAEL), which was found to be a single admin-
data and safety monitoring committee. Data were istration of 3 mg per kilogram infused intrave-
collected by study investigators in New Zealand nously, equivalent to a dose of 1 mg per kilogram

n engl j med 385;6  nejm.org  August 5, 2021 495


The New England Journal of Medicine
Downloaded from nejm.org on July 11, 2023. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

A Intravenous Infusion of NTLA-2001

ApoE opsonization
of LNP in circulation
NTLA-2001

TTR-specific
sgRNA

Complementary
sequence to ApoE protein
TTR gene

Streptococcus Rapid distribution to liver


pyogenes (Spy) through hepatic artery
Cas9 mRNA
Lipid nanoparticle

B NTLA-2001 LNP Uptake in Hepatocytes C Cleavage of DNA at TTR Gene Sequence by Cas9

HEPATOCYTE

CAPILLARY
ApoE protein NUCLEUS

Space of Disse
LDL receptor
HEPATOCYTE sgRNA
Endocytosis of LNP
Cas9

Endosome
Genomic DNA HNH
Complementary
3' 5'
Breakdown of LNP and
release of Cas9 mRNA and
5' PAM 3'
TTR-specific sgRNA into
cytoplasm Noncomplementary
RuvC

Ribonucleoprotein induces Targeted DNA


unwinding of chromosomal cleavage
Translation of Cas9 mRNA DNA, then binds to DNA at
TTR gene, inducing
targeted DNA cleavage 3' 5'

+ 5' 3'
TTR-specific
sgRNA Spy Cas9
endonuclease

Cas9–sgRNA ribonucleoprotein
Scaffold region Endogenous DNA repair
Cas9 through nonhomologous end
sgRNA
Target-specific joining results in introduc-
20-nt sequence 5' tion of indels into TTR gene,
leading to frameshift
mutations that prevent
production of functional TTR
NUCLEUS Cas9–sgRNA ribonucleoprotein protein
enters nucleus

496 n engl j med 385;6 nejm.org August 5, 2021

The New England Journal of Medicine


Downloaded from nejm.org on July 11, 2023. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
CRISPR-C as 9 for Tr ansthyretin Amyloidosis

Figure 1 (facing page). Mechanism of Action of NTLA-


in humans. In accordance with allometric scaling
2001. based on total body-surface area and application
Panel A shows the primary components of NTLA- of a safety factor of 10, the maximum recom-
2001. The carrier system for NTLA-2001 is a lipid mended starting dose of NTLA-2001 for this study
nanoparticle (LNP). The LNP is based on a proprietary was 0.1 mg per kilogram. To mitigate against
ionizable lipid, combined with a phospholipid, a peg­ potential proinflammatory effects of intravenous
ylated lipid (molecular weight of polyethylene glycol,
LNP infusions, patients received glucocorticoid
2000 Da), and cholesterol, formulated in an aqueous
buffer for intravenous administration. The active com- and histamine receptor type 1 and type 2 block-
ponents of NTLA-2001 are a human-optimized mes- ade before infusion.
senger RNA (mRNA) molecule encoding Streptococcus
pyogenes (Spy) Cas9 protein (an approximately Assessments of Clinical Outcomes
4400-nucleotide sequence with a molecular weight
Patients were monitored for assessment of ad-
of approximately 1.5 MDa) and a single guide RNA
(sgRNA) molecule (molecular weight of approximately
verse events and laboratory findings. Serum sam-
35 kDa) specific to the human gene encoding trans- ples were obtained at baseline and at weeks 1, 2,
thyretin (TTR). These components form the cargo of and 4 for analysis of TTR protein levels with a
the LNP for drug administration. After intravenous ad- validated enzyme-linked immunosorbent assay.
ministration of NTLA-2001 and entry into the circula- Evaluations of safety and therapeutic-activity out-
tion, the LNP is opsonized by apolipoprotein E (ApoE)
and transported through the systemic circulation di-
comes are planned for 24 months after NTLA-
rectly into the liver, where it is preferentially distributed. 2001 infusion. In accordance with the require-
Panel B shows transport of the NTLA-2001 LNP into ments of the regulatory authorities that approved
the capillaries of the hepatic sinusoids inside the liver. the current protocol, longer-term safety follow-
As with other clinically approved LNPs,27 NTLA-2001 up is planned under a separate program currently
is then expected to undergo uptake by the low-density
in development.
lipoprotein (LDL) receptor expressed on the surface
of the hepatocytes, followed by endocytosis and endo-
some formation. After breakdown of the LNP and dis- Statistical Analysis
ruption of the endosomal membrane, the active com- Descriptive analyses only were planned. Measure-
ponents (the TTR-specific sgRNA and the mRNA ments of serum TTR protein levels at baseline
encoding Cas9) are released into the cytoplasm. The were compared with those at day 28 and are pre-
Cas9 mRNA molecule is translated through the native
ribosomal process, producing the Cas9 endonuclease
sented as the mean percentage change and range.
enzyme. The TTR-specific sgRNA interacts with the
Cas9 endonuclease, forming a clustered regularly in-
R e sult s
terspaced short palindromic repeats (CRISPR)–Cas9
ribonucleoprotein complex. Panel C shows that the Preclinical Studies of NTLA-2001
Cas9 ribonucleoprotein complex is targeted for nuclear
Preclinical development of NTLA-2001 (Fig. 1)
import and enters the nucleus, where it recognizes
the protospacer-adjacent motif (PAM) on the noncom- included computational modeling and in vitro
plementary DNA strand in TTR. A target-specific studies to ensure on-target gene editing (see the
20-nucleotide sequence at the 5′ end of the sgRNA Supplementary Appendix). In primary human he-
binds to the DNA double helix at the target site, allow- patocytes, NTLA-2001 was highly potent (EC50,
ing the CRISPR-Cas9 complex to unwind the helix and
0.05 to 0.15 nmol per liter; EC90, 0.17 to 0.67
access the target gene. Cas9 undergoes a series of
conformational changes and nuclease domain activa- nmol per liter) and produced saturating levels of
tion (HNH and RuvC domains), resulting in DNA TTR editing (≥93.7%), resulting in 91% or great-
cleavage that is precisely targeted to the TTR sequence, er reductions in TTR mRNA expression and 95%
as defined by the sgRNA complementary sequence. or greater reductions in TTR protein production
Endogenous DNA-repair mechanisms ligate the ends (Fig. 2). Data from next-generation sequencing
of the cut, potentially introducing insertions or dele-
tions of bases (indels). The generation of an indel may
showed that NTLA-2001 induced knockout of TTR.
result in the reduction of functional target-gene mRNA In off-target editing assays, among all the po-
levels as a result of missense or nonsense mutations tential off-target loci identified by Cas-OFFinder,
decreasing the amount of full-length mRNA, ultimate- GUIDE-seq, and SITE-Seq, seven loci were iden-
ly resulting in decreased levels of the target protein. tified as possible editing sites, all of which were
Indels that result in abrogated production of the tar-
located in noncoding regions (Fig. S1 in the Sup-
get protein, in this case TTR, are termed knockout
mutations. plementary Appendix). For each of these sites,
no evidence of off-target editing was found when

n engl j med 385;6  nejm.org  August 5, 2021 497


The New England Journal of Medicine
Downloaded from nejm.org on July 11, 2023. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

tion of serum TTR protein expression at doses


TTR gene editing TTR mRNA expression TTR protein associated with no adverse effects. To evaluate the
production
100
ability of NTLA-2001 to reduce serum expression
90 of TTR protein after a single intravenous infu-
80 sion in humans, we initiated an open-label, single-
70 dose, proof-of-concept study involving patients
Percentage

60
50
who had hATTR amyloidosis with polyneuropathy.
40
30 Patients
20 At one study site (Auckland, New Zealand), three
10
0
patients underwent screening, of whom two were
0.007 0.021 0.063 0.191 0.574 1.724 5.172 15.517 found to be eligible and were enrolled. One par-
sgRNA Concentration (nmol/liter) ticipant had a body weight that was above the
upper limit allowed by the study protocol at that
Figure 2. In Vitro Evaluations of the Potency of NTLA-2001. time. At the other study site (London, United King-
Shown is the relationship between increasing concentrations of sgRNA and dom), four patients underwent screening, all of
the consequent percentages of TTR editing, as well as TTR mRNA expres-
whom were found to be eligible and were enrolled.
sion and TTR protein production in a single lot of primary human hepato-
cytes. The primary indel patterns were a single-nucleotide deletion or in- The patients were 46 to 64 years of age, and four
sertion at the cut site, inducing a frameshift mutation (data not shown). of the six patients were men; the body weight
ranged from 70 to 90 kg. Three patients had a
p.T80A mutation, two a p.S97Y mutation, and one
primary human hepatocytes were treated with a p.H110D mutation. Three patients had received
concentrations of NTLA-2001 up to 3 times as no previous therapy, and three had previously
high as the EC90 (Fig. S2). received diflunisal. All six patients had sensory
Studies in transgenic mice revealed a dose- polyneuropathy in the absence of motor symp-
dependent and durable effect of NTLA-2001. toms (polyneuropathy disability score of 1) and
Editing of TTR reduced circulating serum TTR a New York Heart Association heart failure class
protein levels, which reached a nadir by 4 weeks of I. The level of N-terminal pro–B-type natriuretic
after receipt of the dose and were still maxi- peptide ranged from 50 to 596 ng per liter.
mally suppressed at 12 months of observation.24
After resection of two thirds of the liver and Safety and Side-Effect Profile
subsequent full-liver regeneration, the gene-edit- NTLA-2001 treatment was completed in all the
ing percentage and corresponding protein levels patients without interruption of the infusion. No
were unchanged, findings that supported the protocol-specified stopping events were observed.
permanent nature of the edit (Fig. S7). Adverse events that occurred during or after
Studies in cynomolgus monkeys showed rap- treatment, all of which were mild (grade 1) in
id initial distribution and clearance of the LNP severity, were reported in three of the six pa-
components (Table S3 and Fig. S8). In addition, tients. One patient had an adverse event of spe-
a single dose of Cyn-LNP (the nonhuman primate cial interest (a grade 1 infusion-related reaction;
surrogate of NTLA-2001) at 3 or 6 mg per kilogram see Table S4). No serious adverse events were
was associated with a maximum gene-editing observed. Increased d-dimer levels were ob-
percentage of 73% in whole liver and near-com- served 4 to 24 hours after infusion in five of six
plete (>94%) reduction in serum TTR protein patients; the elevations were lower than those
that was sustained over a period of 12 months observed at the NOAEL dose in nonhuman pri-
(Fig. 3A). Editing of TTR was confirmed by next- mates. The values returned to baseline in all six
generation sequencing analysis of hepatic tissue patients by day 7. Coagulation measures (acti-
(Fig. 3B).25 vated partial thromboplastin time and pro-
Thus, preclinical studies in the mouse and thrombin time) remained within 1.2 times the
cynomolgus monkey showed that a single dose of upper limit of the reference ranges, and fibrino-
NTLA-2001 or its surrogate Cyn-LNP resulted in gen levels and platelet counts remained above
durable TTR editing and near-complete elimina- the lower limit of the reference ranges; liver-

498 n engl j med 385;6  nejm.org  August 5, 2021

The New England Journal of Medicine


Downloaded from nejm.org on July 11, 2023. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
CRISPR-C as 9 for Tr ansthyretin Amyloidosis

function measures (aspartate aminotransferase


A
and alanine aminotransferase levels) remained Control 1.5 mg/kg 3.0 mg/kg 6.0 mg/kg
within normal limits (Fig. S11). (N=3) (N=3) (N=3) (N=3)
120
TTR Protein Reduction 100
To determine the pharmacodynamic effects of

TTR Concentration
(% of baseline)
80
NTLA-2001, concentrations of TTR in serum were
evaluated. Reductions from baseline in the se- 60
rum TTR protein concentration were observed 40
by day 14 and deepened by day 28 (Fig. 4A and
20
4B). At day 28, NTLA-2001 was associated with
mean TTR reductions of 52% in the group that 0
received a dose of 0.1 mg per kilogram and 87% 0 40 80 120 160 200 240 280 320 360

in the group that received 0.3 mg per kilogram Day


(Fig. 4C). The effect was dose-dependent, with
B
greater reductions in TTR concentration among
Indel
patients who received a higher dose of NTLA- Indel Size Sequence Frequency
2001. In addition, the effect of NTLA-2001 was %
reproducible across patients at each dose level, Wild type —
with reductions at day 28 ranging from 47 to 56% +1 98.9
>+1 1.03
in the lower-dose group and from 80 to 96% in
the higher-dose group (Fig. 4A and 4B).
Figure 3. In Vivo Pharmacologic Properties of Cyn-LNP, the Nonhuman
­Primate Surrogate of NTLA-2001.
Discussion Panel A shows mean reductions in the serum TTR protein concentration as
a percentage of the baseline concentration in cynomolgus monkeys (three
We report evidence of CRISPR-Cas9–based in vivo per dose group) that received Cyn-LNP intravenously at doses of 1.5, 3.0,
gene editing in humans. Systemic administration and 6.0 mg of total RNA per kilogram of body weight on day 0 and were fol-
of NTLA-2001 to six patients with hATTR amy- lowed up for 367 days. A control group that received no treatment is shown
for comparison. I bars indicate standard deviations for the three animals in
loidosis with polyneuropathy was associated in each group. Panel B shows the results of next-generation sequencing after
each case with sustained reductions in the serum administration of Cyn-LNP to cynomolgus monkeys. The sgRNA target se-
TTR protein concentration. NTLA-2001 treatment quence is indicated in blue next to the required PAM sequence, shown in
was associated with a dose-dependent effect. At red. [G/A] represents a naturally occurring single-nucleotide polymorphism
day 28, the time at which the drug effect had in the cynomolgus monkeys used in the study. The nucleotide position of
indels relative to the cynomolgus monkey genome (build mf5, chromo-
reached its permanent nadir in preclinical stud- some 18) are +1: 50681549–50681550. The primary indel pattern was a single-
ies, the mean reduction from baseline in serum nucleotide insertion at the cut site, inducing a frameshift mutation. An “N”
TTR protein concentration was 52% in the group at the insertion site indicates a multinucleotide insertion (e.g., AA or AGG),
that received the lower dose (0.1 mg per kilo- which in aggregate constituted 1.03% of all indels. The remaining fraction
gram) and was 87% in the group that received comprised deletions of various lengths.
the higher dose (0.3 mg per kilogram). NTLA-2001
treatment was associated with adverse events of
only mild severity. to note that this study involves a very small num-
These data represent interim results from the ber of patients with limited follow-up to date.
first two dose groups in an ongoing dose-esca- Continued serial measurements of serum TTR
lation study. The results closely follow the pat- concentration in the patients reported here are
tern observed in in vitro data from cell lines and planned to confirm the durability of the effect.
in vivo data on potency in animals, which showed Data from studies of RNA-targeting gene-
a deep and permanent reduction in serum TTR silencing agents have shown that observed re-
protein concentrations with NTLA-2001, thus ductions in serum TTR protein translate into
providing evidence of the potential for in vivo meaningful clinical benefits relative to placebo.
gene editing as a therapeutic strategy for the These agents result in mean reductions from base-
treatment of hATTR amyloidosis. It is important line in serum TTR concentrations of approximate-

n engl j med 385;6  nejm.org  August 5, 2021 499


The New England Journal of Medicine
Downloaded from nejm.org on July 11, 2023. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

A Change in Serum TTR Concentration in Patients Who Received 0.1 mg/kg Figure 4. Reductions from Baseline in Serum TTR
0 ­Protein Concentration after Infusion of NTLA-2001
in Humans.
−10
Panel A shows the percentage change from baseline in
−20
Change from Baseline (%)

total circulating serum TTR protein in the group of pa-


−30 tients who received an NTLA-2001 dose of 0.1 mg per
−40 kilogram. TTR protein was quantified by a validated
−47 enzyme-linked immunosorbent assay method in accor-
−50 −52
−56
dance with regulatory guidelines for biomarker method
−60
validation. Serum samples were measured once, and
−70 each sample was tested in duplicate. In accordance
−80 with good laboratory practice, no retesting was con-
ducted for successful assay runs. For each patient in
−90
the group that received a dose of 0.1 mg per kilogram,
−100 data are shown at postdose days 7, 14, and 28 for per-
0 7 14 21 28
centage reductions from the predose baseline value
Day (mean concentration from three sampling time points).
Panel B shows the percentage change from baseline
B Change in Serum TTR Concentration in Patients Who Received 0.3 mg/kg in total circulating serum TTR protein concentrations
0 in the group of patients who received an NTLA-2001
−10 dose of 0.3 mg per kilogram. The methods and analy-
sis were identical to those described in Panel A. Panel
−20
Change from Baseline (%)

C shows the mean (three per group) percentage reduc-


−30 tion from baseline in total circulating serum TTR pro-
−40 tein at day 28 for both dose groups. The I bars indicate
standard deviations.
−50
−60
−70
−80 −80 serial infusions. On the basis of data in animals,
−84
−90 NTLA-2001 may be able to produce nearly com-
−96
−100 plete and permanent knockdown of TTR expres-
0 7 14 21 28
sion with a single administration. As with cur-
Day rent standard-of-care agents, patients will receive
vitamin A supplementation in order to compen-
C Mean Reduction in Serum TTR Level at Day 28
sate for the loss of TTR, which has a normal
0
physiological role in vitamin A transport.31
−10
The potential for off-target gene editing with
−20
CRISPR-Cas systems has been raised as a concern
Mean Reduction (%)

−30
with regard to the use of these therapies in hu-
−40 mans.32 In primary human hepatocytes, therapeu-
−50 tic concentrations of NTLA-2001 showed no
−60 evidence of previously described off-target muta-
−70 genesis mechanisms.33,34 Computational model-
−80 ing, biochemical cell-free assays, and in vitro
−90 cellular assays were used to identify the most
−100 likely sites in the genome outside of TTR into
0.1 mg/kg 0.3 mg/kg
which NTLA-2001 might introduce off-target
Dose edits. Seven candidate sites for introduction of
indels were confirmed in assays with cell lines
in which supersaturating doses of NTLA-2001
ly 80% and show more favorable clinical effects were used. We assessed the therapeutic index for
in patients in whom lower concentrations of TTR NTLA-2001 in primary human hepatocytes by
protein are achieved. Maintenance of these reduc- determining the frequency of off-target edits at
tions with RNA-targeting agents requires routine concentrations of sgRNA that caused high de-

500 n engl j med 385;6  nejm.org  August 5, 2021

The New England Journal of Medicine


Downloaded from nejm.org on July 11, 2023. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
CRISPR-C as 9 for Tr ansthyretin Amyloidosis

grees of on-target edits and reductions in TTR tional proteins where mutations cause pathologic
protein production. In addition, we performed deficiencies.
long-range sequencing to detect structural vari- The study reported here is ongoing. Dose es-
ants and to determine the risk of unintended calation continues with a goal of producing greater
genotoxicity or oncogenic transformation.35,36 reductions in serum TTR protein than are achieved
Indels and DNA structural variants are natural with available therapies, with anticipated benefi-
outcomes of double-stranded DNA break re- cial effects on disease progression, quality of life,
pair.36 The DNA structural variants induced by and mortality. Data from the initial groups of
CRISPR-Cas9 genome editing were not random patients in this study provide clinical proof of con-
but related to end-joining at the TTR on-target cept for in vivo CRISPR-Cas9–mediated gene
site and are expected to be of low risk. The editing as a therapeutic strategy.
changes detected in the primary human hepato- Supported by Intellia Therapeutics and Regeneron Pharma-
cytes could predict the DNA structural variants ceuticals.
that may occur in vivo. Participants who volunteer Disclosure forms provided by the authors are available with
the full text of this article at NEJM.org.
to receive NTLA-2001 therapy will need to undergo A data sharing statement provided by the authors is available
long-term safety monitoring. with the full text of this article at NEJM.org.
The CRISPR-Cas9 approach used for NTLA-2001 We thank the patients who participated in this study and
their families; New Zealand Clinical Research and Richmond
is modular and has the capacity to be adapted to Pharmacology for contract research assistance; Altasciences,
treat other diseases with simple replacement of Charles River Laboratories, Precision for Medicine, PPD, and
the sgRNA. Indeed, the in vivo gene-editing ap- QPS for assistance with the studies in cynomolgus monkeys,
enzyme-linked immunosorbent assay measurements of serum
proach used in this study is currently being in- transthyretin, and pharmacokinetic and biomarker tests in the
vestigated for use in other diseases. Further first-in-human study; Carri Boiselle, James Butler, David Cooke,
clinical programs involving CRISPR-Cas9–based Tracy DiMezzo, Richard Duncan, Eva Essig, Noah Gardner, Bo
Han, Denise Hernandez, Tracy Jennings, Kellie Kolb, Rebecca
gene-editing strategies are planned by many in- Lescarbeau, Reynald Lescarbeau, Nishit Patel, Austin Ricker,
vestigators for a wide range of diseases; these Joseph Rissman, Matthew Roy, Philipp Schneggenburger, Palak
programs may make use of the potential not Sharma, and Samantha Soukamneuth from Intellia Therapeu-
tics for their dedicated investment in the development of NTLA-
only to knock out expression of harmful protein 2001; and Ben Caldwell of Arc, a division of Spirit Medical Com-
products but also to insert genes to produce func- munications Group, for providing medical writing services.

References
1. Marcoux J, Mangione PP, Porcari R, 7. Maurer MS, Bokhari S, Damy T, et al. 13. Adams D, Polydefkis M, González-
et al. A novel mechano-enzymatic cleav- Expert consensus recommendations for Duarte A, et al. Long-term safety and
age mechanism underlies transthyretin the suspicion and diagnosis of transthyre- efficacy of patisiran for hereditary trans-
amyloidogenesis. EMBO Mol Med 2015;​7:​ tin cardiac amyloidosis. Circ Heart Fail thyretin-mediated amyloidosis with poly-
1337-49. 2019;​12(9):​e006075. neuropathy: 12-month results of an open-
2. Gertz MA, Benson MD, Dyck PJ, et al. 8. Merlini G, Coelho T, Waddington label extension study. Lancet Neurol 2021;​
Diagnosis, prognosis, and therapy of Cruz M, Li H, Stewart M, Ebede B. Evalu- 20:​49-59.
transthyretin amyloidosis. J Am Coll Car- ation of mortality during long-term treat- 14. Solomon SD, Adams D, Kristen A, et al.
diol 2015;​66:​2451-66. ment with tafamidis for transthyretin Effects of patisiran, an RNA interference
3. Ando Y, Coelho T, Berk JL, et al. amyloidosis with polyneuropathy: clinical therapeutic, on cardiac parameters in pa-
Guideline of transthyretin-related heredi- trial results up to 8.5 years. Neurol Ther tients with hereditary transthyretin-medi-
tary amyloidosis for clinicians. Orphanet 2020;​9:​105-15. ated amyloidosis. Circulation 2019;​ 139:​
J Rare Dis 2013;​8:​31. 9. Maurer MS, Schwartz JH, Gundapa- 431-43.
4. Hawkins PN, Ando Y, Dispenzeri A, neni B, et al. Tafamidis treatment for pa- 15. Urits I, Swanson D, Swett MC, et al.
Gonzalez-Duarte A, Adams D, Suhr OB. tients with transthyretin amyloid cardio- A review of patisiran (ONPATTRO) for the
Evolving landscape in the management of myopathy. N Engl J Med 2018;​379:​1007-16. treatment of polyneuropathy in people
transthyretin amyloidosis. Ann Med 2015;​ 10. Berk JL, Suhr OB, Obici L, et al. Re- with hereditary transthyretin amyloido-
47:​625-38. purposing diflunisal for familial amyloid sis. Neurol Ther 2020;​9:​301-15.
5. Schmidt HH, Waddington-Cruz M, polyneuropathy: a randomized clinical 16. Lozeron P, Théaudin M, Mincheva Z,
Botteman MF, et al. Estimating the global trial. JAMA 2013;​310:​2658-67. Ducot B, Lacroix C, Adams D. Effect on
prevalence of transthyretin familial amy- 11. Benson MD, Waddington-Cruz M, disability and safety of tafamidis in late
loid polyneuropathy. Muscle Nerve 2018;​ Berk JL, et al. Inotersen treatment for pa- onset of Met30 transthyretin familial
57:​829-37. tients with hereditary transthyretin amy- amyloid polyneuropathy. Eur J Neurol
6. Dohrn MF, Ihne S, Hegenbart U, et al. loidosis. N Engl J Med 2018;​379:​22-31. 2013;​20:​1539-45.
Targeting transthyretin — mechanism- 12. Adams D, Gonzalez-Duarte A, 17. Gertz MA, Scheinberg M, Wadding-
based treatment approaches and future O’Riordan WD, et al. Patisiran, an RNAi ton-Cruz M, et al. Inotersen for the treat-
perspectives in hereditary amyloidosis. therapeutic, for hereditary transthyretin ment of adults with polyneuropathy
J Neurochem 2021;​156:​802-18. amyloidosis. N Engl J Med 2018;​379:​11-21. caused by hereditary transthyretin-medi-

n engl j med 385;6  nejm.org  August 5, 2021 501


The New England Journal of Medicine
Downloaded from nejm.org on July 11, 2023. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
CRISPR-C as 9 for Tr ansthyretin Amyloidosis

ated amyloidosis. Expert Rev Clin Phar- 24. Finn JD, Smith AR, Patel MC, et al. 30. Cameron P, Fuller CK, Donohoue PD,
macol 2019;​12:​701-11. A single administration of CRISPR/Cas9 et al. Mapping the genomic landscape of
18. Jinek M, Chylinski K, Fonfara I, Hauer lipid nanoparticles achieves robust and CRISPR-Cas9 cleavage. Nat Methods 2017;​
M, Doudna JA, Charpentier E. A program- persistent in vivo genome editing. Cell 14:​600-6.
mable dual-RNA-guided DNA endonucle- Rep 2018;​22:​2227-35. 31. Müller ML, Butler J, Heidecker B.
ase in adaptive bacterial immunity. Science 25. Wood K, Pink M, Seitzer J, et al. De- Emerging therapies in transthyretin am-
2012;​337:​816-21. velopment of NTLA-2001, a CRISPR/Cas9 yloidosis — a new wave of hope after
19. Hsu PD, Lander ES, Zhang F. Develop- genome editing therapeutic for the treat- years of stagnancy? Eur J Heart Fail 2020;​
ment and applications of CRISPR-Cas9 for ment of ATTR. Presented at the Second 22:​39-53.
genome engineering. Cell 2014;​157:​1262- European Congress for ATTR Amyloido- 32. Lin Y, Cradick TJ, Brown MT, et al.
78. sis, Berlin, September 1–3, 2019. CRISPR/Cas9 systems have off-target ac-
20. Dasgupta I, Flotte TR, Keeler AM. 26. Sabnis S, Kumarasinghe ES, Salerno tivity with insertions or deletions between
CRISPR/Cas-dependent and nuclease-free T, et al. A novel amino lipid series for target DNA and guide RNA sequences.
in vivo therapeutic gene editing. Hum mRNA delivery: improved endosomal es- Nucleic Acids Res 2014;​42:​7473-85.
Gene Ther 2021;​32:​275-93. cape and sustained pharmacology and 33. Haapaniemi E, Botla S, Persson J,
21. Li H, Yang Y, Hong W, Huang M, Wu safety in non-human primates. Mol Ther Schmierer B, Taipale J. CRISPR-Cas9 ge-
M, Zhao X. Applications of genome edit- 2018;​26:​1509-19. nome editing induces a p53-mediated DNA
ing technology in the targeted therapy of 27. Akinc A, Maier MA, Manoharan M, damage response. Nat Med 2018;​24:​927-30.
human diseases: mechanisms, advances et al. The Onpattro story and the clinical 34. Enache OM, Rendo V, Abdusamad M,
and prospects. Signal Transduct Target translation of nanomedicines containing et al. Cas9 activates the p53 pathway and
Ther 2020;​5:​1. nucleic acid-based drugs. Nat Nanotech- selects for p53-inactivating mutations. Nat
22. Power DM, Elias NP, Richardson SJ, nol 2019;​14:​1084-7. Genet 2020;​52:​662-8.
Mendes J, Soares CM, Santos CR. Evolu- 28. Bae S, Park J, Kim JS. Cas-OFFinder: 35. Vilenchik MM, Knudson AG. Endoge-
tion of the thyroid hormone-binding pro- a fast and versatile algorithm that search- nous DNA double-strand breaks: produc-
tein, transthyretin. Gen Comp Endocrinol es for potential off-target sites of Cas9 tion, fidelity of repair, and induction of
2000;​119:​241-55. RNA-guided endonucleases. Bioinformat- cancer. Proc Natl Acad Sci U S A 2003;​100:​
23. van Bennekum AM, Wei S, Gamble ics 2014;​30:​1473-5. 12871-6.
MV, et al. Biochemical basis for depressed 29. Tsai SQ, Zheng Z, Nguyen NT, et al. 36. Varga T, Aplan PD. Chromosomal ab-
serum retinol levels in transthyretin-defi- GUIDE-seq enables genome-wide profil- errations induced by double strand DNA
cient mice. J Biol Chem 2001;​276:​1107- ing of off-target cleavage by CRISPR-Cas breaks. DNA Repair (Amst) 2005;​4:​1038-46.
13. nucleases. Nat Biotechnol 2015;​33:​187-97. Copyright © 2021 Massachusetts Medical Society.

502 n engl j med 385;6  nejm.org  August 5, 2021

The New England Journal of Medicine


Downloaded from nejm.org on July 11, 2023. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.

You might also like